Virosome-formulated Plasmodium falciparum AMA-1 & CSP derived peptides as malaria vaccine: randomized phase 1b trial in semi-immune adults & children

Patrick Georges Cech, Thomas Aebi, Mwanajaa Shomari Abdallah, Maxmillian Mpina, Ester Barnabas Machunda, Nicole Westerfeld, Sabine Alexandra Stoffel, Rinaldo Zurbriggen, Gerd Pluschke, Marcel Tanner, Claudia Daubenberger, Blaise Genton, Salim Abdulla, Patrick Georges Cech, Thomas Aebi, Mwanajaa Shomari Abdallah, Maxmillian Mpina, Ester Barnabas Machunda, Nicole Westerfeld, Sabine Alexandra Stoffel, Rinaldo Zurbriggen, Gerd Pluschke, Marcel Tanner, Claudia Daubenberger, Blaise Genton, Salim Abdulla

Abstract

Background: This trial was conducted to evaluate the safety and immunogenicity of two virosome formulated malaria peptidomimetics derived from Plasmodium falciparum AMA-1 and CSP in malaria semi-immune adults and children.

Methods: The design was a prospective randomized, double-blind, controlled, age-deescalating study with two immunizations. 10 adults and 40 children (aged 5-9 years) living in a malaria endemic area were immunized with PEV3B or virosomal influenza vaccine Inflexal®V on day 0 and 90.

Results: No serious or severe adverse events (AEs) related to the vaccines were observed. The only local solicited AE reported was pain at injection site, which affected more children in the Inflexal®V group compared to the PEV3B group (p = 0.014). In the PEV3B group, IgG ELISA endpoint titers specific for the AMA-1 and CSP peptide antigens were significantly higher for most time points compared to the Inflexal®V control group. Across all time points after first immunization the average ratio of endpoint titers to baseline values in PEV3B subjects ranged from 4 to 15 in adults and from 4 to 66 in children. As an exploratory outcome, we found that the incidence rate of clinical malaria episodes in children vaccinees was half the rate of the control children between study days 30 and 365 (0.0035 episodes per day at risk for PEV3B vs. 0.0069 for Inflexal®V; RR = 0.50 [95%-CI: 0.29-0.88], p = 0.02).

Conclusion: These findings provide a strong basis for the further development of multivalent virosomal malaria peptide vaccines.

Trial registration: ClinicalTrials.gov NCT00513669.

Keywords: Tanzania; efficacy; immunogenicity; malaria; peptide; safety; vaccine; virosomes.

Conflict of interest statement

Competing Interests: NW, SAS and RZ were employees of Pevion Biotech Ltd. during the trial. RZ and GP are named as inventors in patents covering the vaccines tested. By the end of 2007, Mymetics S.A., Lausanne, Switzerland took over the responsibility of this project from Pevion Biotech Ltd. PC's curent affiliation is with F. Hoffmann-La Roche Ltd., Basel, Switzerland and RZ's current affiliation is with Lonza Ltd., Visp, Switzerland. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Study flow chart.
Figure 1. Study flow chart.
Figure 2. Development of anti-AMA49-C1 and anti-UK-39…
Figure 2. Development of anti-AMA49-C1 and anti-UK-39 IgG ELISA endpoint titres in adults and children immunized with PEV3B or Inflexal®V.
Bars indicate 95% confidence intervals of the geometric mean (no bars are shown for adult Inflexal®V, n = 2). Participants were immunized on days 0 and 90 (see arrows).
Figure 3. Box-plots of the logarithms of…
Figure 3. Box-plots of the logarithms of the index of response (ratios of anti-AMA49-C1 and anti-UK-39 IgG ELISA endpoint to baseline titers) for samples taken at day 30, 90, 120, 180, and 365.
AP: adults PEV3B, AI: adults Inflexal®V, CP: children PEV3B, CI: children Inflexal®V. At all time points the logarithm of the index of response of AP and CP groups were significantly >0 (index of response >1, p<0.05). This was never observed for the AI and CI groups. Symbols indicate the level of significance for differences in the index of response between PEV3B and Inflexal®V groups within adults and children subjects (exact Wilcoxon test, *: p<0.05, **: p<0.01, ***: p<0.001).
Figure 4. Attack rates of first or…
Figure 4. Attack rates of first or only clinical malaria episodes in vaccinated children.
Kaplan-Meyer failure functions are shown for the intervals from 30 days after first vaccination (day 30) until the end of the study (day 365), from 30 days after first vaccination until second vaccination (day 90), and from 30 days after second vaccination (day 120) until study end (day 365). P-values for differences in failure functions between treatment groups are given (log-rank test). Note: the failure function in the third panel is not identical with the corresponding part of the failure function in the first panel, because of the occurrence of multiple events in the same individual during the course of the study.
Figure 5. Comparison of the integrated antibody…
Figure 5. Comparison of the integrated antibody responses against AMA49-C1 and UK-39 with the detection of malaria parasitaemia in PEV3B children during time periods Day 0–30, Day 0–120 and Day 0–365.
ΔAUC was calculated as the area under the log (antibody titer)-curve above the baseline antibody titer. ΔAUC values were compared between those PEV3B children who did not have positive smears for P. falciparum and those PEV3B children who were tested positive for parasites using Wilcoxon's test. The numbers (in brackets) in the bars indicate the number of PEV3B in each of the groups compared.

References

    1. The malERA Consultative Group on Vaccines. A Research Agenda for Malaria Eradication: Vaccines. PLoS Medicine. 2011;8:e1000398.
    1. Pierce SK, Miller LH. World Malaria Day 2009: What Malaria Knows about the Immune System That Immunologists Still Do Not. Journal of Immunology. 2009;182:5171–5177.
    1. Crompton PD, Pierce SK, Miller LH. Advances and challenges in malaria vaccine development. Journal of Clinical Investigation. 2010;120:4168–4178.
    1. Good MF. Vaccine-induced immunity to malaria parasites and the need for novel strategies. Trends in Parasitology. 2005;21:29–34.
    1. Kappe SHI, Buscaglia CA, Nussenzweig V. Plasmodium sporozoite molecular cell biology. Annual Review of Cell and Developmental Biology. 2004;20:29–59.
    1. Coppi A, Tewari R, Bishop JR, Bennett BL, Lawrence R, et al. Heparan sulfate proteoglycans provide a signal to Plasmodium sporozoites to stop migrating and productively invade host cells. Cell Host & Microbe. 2007;2:316–327.
    1. Okitsu SL, Kienzl U, Moehle K, Silvie O, Peduzzi E, et al. Structure-activity-based design of a synthetic malaria peptide eliciting sporozoite inhibitory antibodies in a virosomal formulation. Chemistry & Biology. 2007;14:577–587.
    1. Zavala F, Tam JP, Hollingdale MR, Cochrane AH, Quakyi I, et al. Rationale for Development of a Synthetic Vaccine against Plasmodium-Falciparum Malaria. Science. 1985;228:1436–1440.
    1. Ghasparian A, Moehle K, Linden A, Robinson JA. Chemical Communications; 2006. Crystal structure of an NPNA-repeat motif from the circumsporozoite protein of the malaria parasite Plasmodium falciparum. pp. 174–176.
    1. Okitsu SL, Silvie O, Westerfeld N, Curcic M, Kammer AR, et al. A Virosomal Malaria Peptide Vaccine Elicits a Long-Lasting Sporozoite-Inhibitory Antibody Response in a Phase 1a Clinical Trial. Plos One. 2007;2
    1. Anders RF, Adda CG, Foley M, Norton RS. Recombinant protein vaccines against the asexual blood stages of Plasmodium falciparum. Human Vaccines. 2010;6:39–53.
    1. Cowman AF, Crabb BS. Invasion of red blood cells by malaria parasites. Cell. 2006;124:755–766.
    1. Girard MP, Reed ZH, Friede M, Kieny MP. A review of human vaccine research and development: Malaria. Vaccine. 2007;25:1567–1580.
    1. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, et al. A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. Journal of Biological Chemistry. 2004;279:9490–9496.
    1. Remarque EJ, Faber BW, Kocken CHM, Thomas AW. Apical membrane antigen 1: a malaria vaccine candidate in review. Trends in Parasitology. 2008;24:74–84.
    1. Treeck M, Tamborrini M, Daubenberger CA, Gilberger TW, Voss TS. Caught in action: mechanistic insights into antibody-mediated inhibition of Plasmodium merozoite invasion. Trends in Parasitology. 2009;25:494–497.
    1. Mueller MS, Renard A, Boato F, Vogel D, Naegeli M, et al. Induction of parasite growth-inhibitory antibodies by a virosomal formulation of a peptidomimetic of loop I from domain III of Plasmodium falciparum apical membrane antigen 1. Infection and Immunity. 2003;71:4749–4758.
    1. Zurbriggen R. Immuno stimulating reconstituted influenza virosomes. Vaccine. 2003;21:921–924.
    1. Poltl-Frank F, Zurbriggen R, Helg A, Stuart F, Robinson J, et al. Use of reconstituted influenza virus virosomes as an immunopotentiating delivery system for a peptide-based vaccine. Clinical and Experimental Immunology. 1999;117:496–503.
    1. Peduzzi E, Westerfeld N, Zurbriggen R, Pluschke G, Daubenberger CA. Contribution of influenza immunity and virosomal-formulated synthetic peptide to cellular immune responses in a phase I subunit malaria vaccine trial. Clinical Immunology. 2008;127:188–197.
    1. Tamborrini M, Mueller MS, Stoffel SA, Westerfeld N, Vogel D, et al. Design and pre-clinical profiling of a Plasmodium falciparum MSP-3 derived component for a multi-valent virosomal malaria vaccine. Malaria Journal. 2009;8
    1. Pancharoen C, Mekmullica J, Thisyakorn U, Kasempimolporn S, Wilde H, et al. Reduced-dose intradermal vaccination against hepatitis A with an aluminum-free vaccine is immunogenic and can lower costs. Clinical Infectious Diseases. 2005;41:1537–1540.
    1. Clarke PD, Adams P, Ibanez R, Herzog C. Rate, intensity, and duration of local reactions to a virosome-adjuvanted vs. an aluminium-adsorbed hepatitis A vaccine in UK travellers. Travel Med Infect Dis. 2006;4:313–318.
    1. Genton B, Pluschke G, Degen L, Kammer AR, Westerfeld N, et al. A Randomized Placebo-Controlled Phase Ia Malaria Vaccine Trial of Two Virosome-Formulated Synthetic Peptides in Healthy Adult Volunteers. Plos One. 2007;2
    1. Thompson FM, Porter DW, Okitsu SL, Westerfeld N, Vogel D, et al. Evidence of Blood Stage Efficacy with a Virosomal Malaria Vaccine in a Phase IIa Clinical Trial. Plos One. 2008;3
    1. Kammer AR, Amacker M, Rasi S, Westerfeld N, Gremion C, et al. A new and versatile virosomal antigen delivery system to induce cellular and humoral immune responses. Vaccine. 2007;25:7065–7074.
    1. Missinou MA, Lell B, Kremsner PG. Uncommon asymptomatic Plasmodium falciparum infections in Gabonese children. Clinical Infectious Diseases. 2003;36:1198–1202.
    1. Moorthy VS, Reed ZH, Smith PG. MALVAC 2008: Measures of efficacy of malaria vaccines in phase 2b and phase 3 trials - Scientific, regulatory and public health perspectives. Vaccine. 2009;27:624–628.
    1. Sacarlal J, Aide P, Aponte JJ, Renom M, Leach A, et al. Long-Term Safety and Efficacy of the RTS,S/AS02A Malaria Vaccine in Mozambican Children. Journal of Infectious Diseases. 2009;200:329–336.
    1. Pink JR, Kieny M-P. 4th meeting on Novel Adjuvants Currently in/close to Human Clinical Testing World Health Organization – organisation Mondiale de la Sante Fondation Merieux, Annecy, France, 23-25, June 2003. Vaccine. 2004;22:2097–2102.
    1. Saul A, Lawrence G, Allworth A, Elliott S, Anderson K, et al. A human phase 1 vaccine clinical trial of the Plasmodium falciparum malaria vaccine candidate apical membrane antigen 1 in Montanide ISA720 adjuvant. Vaccine. 2005;23:3076–3083.
    1. Spring MD, Cummings JF, Ockenhouse CF, Dutta S, Reidler R, et al. Phase 1/2a Study of the Malaria Vaccine Candidate Apical Membrane Antigen-1 (AMA-1) Administered in Adjuvant System AS01B or AS02A. Plos One. 2009;4
    1. Wu YM, Ellis RD, Shaffer D, Fontes E, Malkin EM, et al. Phase 1 Trial of Malaria Transmission Blocking Vaccine Candidates Pfs25 and Pvs25 Formulated with Montanide ISA 51. Plos One. 2008;3
    1. Casares S, Brumeanu T-D, Richie TL. The RTS,S malaria vaccine. Vaccine. 2010;28:4880–4894.
    1. Alonso PL, Sacarlal J, Aponte JJ, Leach A, Macete E, et al. Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial. Lancet. 2004;364:1411–1420.
    1. Greenwood B. Immunological correlates of protection for the RTS,S candidate malaria vaccine. Lancet Infectious Diseases. 2011;11:75–76.
    1. James S, Moehle K, Renard A, Mueller MS, Vogel D, et al. Synthesis, solution structure and immune recognition of an epidermal growth factor-like domain from Plasmodium falciparum merozoite surface protein-1. Chembiochem. 2006;7:1943–1950.
    1. Okitsu SL, Boato F, Mueller MS, Li DB, Vogel D, et al. Antibodies elicited by a virosomally formulated Plasmodium falciparum serine repeat antigen-5 derived peptide detect the processed 47 kDa fragment both in sporozoites and merozoites. Peptides. 2007;28:2051–2060.

Source: PubMed

3
Abonner